Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies
Open Access

Effectiveness of Rhenium(I)-diselenoether Low Doses in a Triple-negative Breast Cancer Chicken Embryo Model

PHILIPPE COLLERY, CHLOE PRUNIER, EMILIEN DOSDA, JEAN VIALLET, ADHIKESAVAN HARIKRISHNAN, VIJAY VEENA and DIDIER DESMAELE
Anticancer Research March 2024, 44 (3) 941-951; DOI: https://doi.org/10.21873/anticanres.16889
PHILIPPE COLLERY
1Société de Coordination de Recherches Thérapeutiques, Algajola, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: philippe.collery{at}gmail.com
CHLOE PRUNIER
2Inovotion, La Tronche, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
EMILIEN DOSDA
2Inovotion, La Tronche, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JEAN VIALLET
2Inovotion, La Tronche, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ADHIKESAVAN HARIKRISHNAN
3Department of Chemistry, School of Arts and Science, Vinayaka Missions Research Foundation (DU), Chennai, India;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
VIJAY VEENA
4School of Allied Healthcare and Sciences, Jain University, Bangalore, India;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: btveenavijaykumar{at}gmail.com
DIDIER DESMAELE
5Institut Galien, Orsay, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Rhenium(I)-diselenoether (Re-diSe) is a promising anticancer agent composed of one rhenium and two selenium atoms. Its effectiveness was established in inhibiting cancer cells while maintaining low toxicity toward normal cells at a 5 μM dose for 120 hours in MDA-MB-231 cells. In MDA-MB-231 breast tumor-bearing mice, anti-tumor and anti-metastatic effects were observed at a 10 mg/kg dose. However, contradictory results were observed in the 4T1 breast cancer model, where a dose of 60 mg/kg had a pro-tumor effect. To address these discrepancies, the efficacy of Re-diSe at the effective 10 mg/kg dose was validated in a transplanted MDA-MB-231 breast tumor model using the chicken chorioallantoic membrane assay. Materials and Methods: MDA-MB-231 cancer cells were xenografted onto the chicken chorioallantoic membrane (CAM), and daily drug administration was carried out for nine days at doses of 0.1, 1, and 10 mg/kg. At the study’s conclusion, a standard histological analysis was conducted. Results: The low dose of 0.1 mg/kg showed a significant reduction in tumor weights compared to controls. The 1 mg/kg dose resulted in an increased inflammation score but did not induce a significant difference in tumor weights compared to the 0.1 mg/kg dose. Notably, at the 10 mg/kg dose, six out of 11 treated embryos displayed no visible signs of tumors. These tumors exhibited extensive tumor necrosis and significant infiltration by inflammatory cells. Conclusion: In this particular model, the anticancer efficacy of Re-diSe was achieved at the low dose of 0.1 mg/kg. The higher dose of 10 mg/kg, while eliminating visible tumors, might have immune-mediated effects, as indicated by substantial tumor necrosis and infiltration by inflammatory cells. Overall, this study successfully demonstrated the effectiveness of Re-diSe as an anticancer agent.

Key Words:
  • Rhenium
  • selenium
  • breast cancer
  • inflammation
  • dose-effect
  • chicken embryo model

The rhenium(I)-diselenoether (Re-diSe) is a metal-based drug that displays a fac-[Re(CO)3]-core tightly bonded to 3,7-diselenanonanedioic acid disodium salt ligand stabilizing the metal and allowing water solubility (1). It was found to selectively inhibit the growth of cancer cells versus normal cells, decreasing selectively the production of reactive oxygen species (ROS), transforming growth factor-beta (TGF-β), vascular epidermal growth factor A (VEGF-A) and insulin growth factor 1 (IGF1) by the cancer cells (2), even when the culture medium of the cells was enriched in inflammatory cytokines (3). Its antitumor activity was demonstrated in an experimental model of triple-negative breast cancer (TNBC) with MDA-MB-231 orthotopically transplanted tumor-bearing nude mice, after an oral administration of 10 mg/kg for 28 days, both on the primary tumors and the metastases (4). A significant antitumor effect was confirmed in the same model after a daily intraperitoneal administration for 28 days, without differences between the doses of 10 and 40 mg/kg, while higher doses induced a toxicity without improving efficiency (5). Further experimental studies showed controversial results. A pro-tumor effect was observed at daily oral doses of 5 or 10 mg/kg Re-diSe for six weeks in the MDA-MB-231 model, but in immunodeficient mice treated with a total-body irradiation prior to the transplantation of the tumor cells, suppressing thus their immune defense. High daily oral doses of 60 mg/kg Re-diSe induced a pro-tumor effect in 4T1- transplanted tumors in non-immune deficient mice, while no effect on the tumor growth was noted at lower doses of 0.1, 1, and 10 mg/kg. It was concluded that the choice of the model was crucial to determine the efficacy of Re-diSe. Therefore, we wanted to confirm the efficacy of the Re-diSe drug in MDA-MB-231 tumor-bearing mice at the dose of 10 mg/kg, which already showed an anticancer activity in this model of triple-negative breast cancer, and at lower doses.

The chicken chorioallantoic membrane assay has the advantage to rapidly develop within 8 days measurable tumors and represents an alternative to experiments in tumor-transplanted nude mice (6-8). We used MDA-MB-231 breast cancer cells xenografted on the chicken chorioallantoic membrane (CAM) to assess the efficacy of the Re-diSe drug. To evaluate a dose-dependent effect, Re-diSe was administered for nine days at three different doses: 0.1, 1, and 10 mg/kg from the embryonic developmental day 10 (EDD10) to EDD18; the embryos were sacrificed at EDD19. The efficacy was evaluated through the measurement of the tumor weights and by a standard histological analysis performed on fixed tumors at the end of the experiment. The hematoxylin and eosin histological analysis allowed to study the tumor cells, the infiltration by inflammatory cells, and the tumor necrosis. The toxicity was evaluated by comparing the number of dead embryos in Re-diSe-treated and control (vehicle) embryos.

Materials and Methods

The chicken embryo CAM assay was performed by INOVOTION SAS (La Tronche, France). No ethical approval is required for scientific experimentation using oviparous embryos (decree number 2013-118, February 1, 2013; art. R214-88) according to French legislation.

Synthesis. Rhenium(I)diseleno-ether (Re-diSe) was synthesized by ligand exchange from pentacarbonylchlororhenium with 3,7-diselenanonanedioic acid. The obtained dicarboxylic acid rhenium complex was converted into its water soluble bis sodium salt upon treatment with sodium carbonate as reported earlier (1).

Design. Fertilized white Leghorn eggs (Couvoir Hubert, Guilberville, France) were incubated at 37.5°C with 50% relative humidity for nine days. At EDD9, eggs were randomized into groups and the CAM was dropped down by drilling a small hole through the eggshell into the air sac, and a 1 cm2 window was cut in the eggshell above the CAM. Then, 1×106 MDA-MB-231 cells (from ATCC, Manassas, VA, USA) in 50 μl of culture media were inoculated onto the egg’s CAM.

Treatments. From EDD10 to EDD18, eggs received either vehicle (water, n=16), Re-diSe [1] (0.1 mg/kg, n=17), Re-diSe [2] (1 mg/kg, n=17) and Re-diSe [3] (10 mg/kg, n=19) every day.

Efficacy. Evaluation of the efficacy on the primary tumors: at EDD19, tumors which were attached on the upper portion of the CAM were removed, washed with PBS buffer, and then fixed in 4% PFA for 48 h. Tumors were carefully cut away from normal CAM tissue and weighed.

Histological observations. PFA-fixed tumors were trimmed and embedded in paraffin cassettes (n=11 per group). Paraffin sections were cut (4 μm thick) and put on glass slides. Slides were stained with hematoxylin & eosin for general histopathological evaluation.

Non-overlapping fields were graded by a semi-quantitative scoring system for the presence of pathological changes (n=10 fields per tumor).

Pictures were taken using a microscope (Olympus BX60, serial NO.7D04032, Rungis, France) at objective magnification X1.25 and X10, and microscope’s camera (Olympus DP73, serial NO. 0H05504).

It has been demonstrated that the tumor inflammatory cell infiltrate and the tumor necrosis could be assessed on hematoxylin and eosin-stained sections (9-11). The intensity of overall inflammatory cell reaction, numbers of neutrophilic and eosinophilic granulocytes, lymphoid cells and macrophages is scored using this method (12).

Inflammation was scored from 0 to 3 with 0: scant or absent stroma cells, 1: stroma cells obviously present but markedly less than tumor cells, 2: stroma cells roughly equal to tumor cells and 3: predominantly stroma cells.

Tumor necrosis was scored from 0 to 5 with 0: no necrosis, 1: small foci of necrosis, 2: necrosis in <50% of the field, 3: necrosis in 50-75% of the field, 4: necrosis in 75-90% of the field and 5: necrosis in >90% of the field.

Statistical analysis. A one-way ANOVA analysis with Tukey’s multiple comparison post-test was performed on the dataset. For all analyses, statistically significant differences between groups are indicated on the graphs using stars as follows: - No star: No statistically significantly different (p-value >0.05); - One star (*): 0.05 ≥ p-value >0.01; - Two stars (**): 0.01 ≥ p-value >0.001; - Three stars (***): 0.001 ≥ p-value ≥0.0001; - Four stars (****): p-value ≤0.0001.

Results

Efficacy. A significant decrease of the tumor weights was observed at the dose of 0.1 mg/kg Re-diSe versus controls, from 25.74±1.95 mg in controls to 16.73±1.98 mg. The tumor weights in the group treated with 1 mg/kg (15.38±1.92 mg) did not significantly differ compared to those with the 0.1 mg/kg doses. No visible tumors were observed in 6/11 embryos treated with 10 mg/kg. In the other 5/11 embryos treated with this dose, the tumor weights significantly decreased to 8.74±1.81 mg by comparison with controls (Figure 1). The remarkable visual difference of the CAM between controls and the group treated at the dose of 10 mg/kg is shown in Figure 2. Table I emphasizes the absence of visible tumor in embryos treated with 10 mg/kg.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Average tumor weights (±SEM; n=11-14 embryos per group and n=5 in Re-diSe [3]). Human MDA-MB-231 breast cancer cells were grafted onto the chorioallantoic membrane and treated daily with vehicle (Neg. Ctrl.), Re-diSe at 0.1 mg/kg (Re-diSe [1]), 1 mg/kg (Re-diSe [2]) and 10 mg/kg (Re-diSe [3]). Tumors were collected 10 days after the graft, fixed in PFA and weighed. Only 5/11 tumors were visible in Re-diSe [3] and therefore included in the statistical analysis and shown in this figure. (**): 0.01 ≥ p-value >0.001, (***): 0.001 ≥ p-value ≥0.0001.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Illustrative pictures of the typical aspect of an MDA-MB-231 tumor grown on the CAM in the Neg. Ctrl. group (A) and the inflammatory membrane without visible tumor in 6/11 Re-diSe [3] treated embryos (B).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Absence of visible tumor in 6/11 Re-diSe [3] treated embryos.

Toxicity. There is a known lethal effect of the transplantation of the tumor cells on the embryos. The number of deaths was increased in all treated groups versus controls, but not significantly. The results are shown in Figure 3.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Kaplan–Meier curve summarizing the embryo survival of Re-diSe-treated eggs. Sixteen to nineteen embryos per group were grafted with MDA-MB-231 cancer cells at developmental day 9 (EDD9). Viability of the embryos was examined daily, and the death index recorded up to EDD19. Log-rank (Mantel–Cox) statistical test was used to analyze the results. p-Values are shown on the right part of the graph.

Histological evaluation. In controls, tumors consisted of a central, localized, nodular dense mass with round to spindle shape, large neoplastic cells with a very high mitotic activity and with clear cell borders. The central core was surrounded by a very loose network of neoplastic cells. High grade of nuclear pleomorphism and many multi-nucleated giant cells were observed in the neoplastic cells population. The score of inflammation cells was mild.

In the group treated with 10 mg/kg Re-diSe, 6 of the 11 samples showed only embryo membranes with very few surviving neoplastic cells and some necrotic material. The physiological aspect of the CAM was altered in these samples (thickening, absence of vascularization and whitening of the membrane at the grafting site).

The score of inflammation did not vary between controls (1.455±0.157) and embryos treated with 0.1 mg/kg Re-diSe (1.545±0.157), but significantly increased upon treatment with 1 mg/kg Re-diSe (2.091±0.162), and 10 mg/kg Re-diSe (2.091±0.090).

The score of tumor necrosis increased from 2±0.233 in controls to 2.182±0.181 at the dose of 0.1 mg/kg (N.S), and to 2.636±0.203 at the dose of 1 mg/kg (N.S). The tumor necrosis was highly significantly increased in the group treated with 10 mg/kg (4.182±0.325) and significantly compared with the two other treated groups. The results are shown in Figure 4.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Histological analysis of tumors treated with Re-diSe. Tumors fixed in PFA4% were included in paraffin blocks, sliced, and stained with hematoxylin and eosin. The average score (±SEM) for inflammation (A) and necrosis (B) was assessed by a histopathologist using a semi-quantitative scoring technique (see Materials and Methods). Ordinary one-way ANOVA with Tukey’s multiple comparison post-test was used to analyze the results. n=11 tumors per group. (*): 0.05 ≥ p-value >0.01; (***): 0.001 ≥ p-value ≥ 0.0001; (****): p-value ≤0.0001.

Microscopical pictures. Images of the microscopic observations in Figure 5 illustrate the remarkable rarefaction of the tumors cells, the infiltration by inflammatory cells and the huge necrosis of the tumors at the dose of 10 mg/kg with intermediate results at the lower doses. The CAM was altered at the dose of 10 mg/kg and appeared inflammatory.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Illustrative pictures of hematoxylin and eosin stained tumors. Tumors fixed in PFA4% were included in paraffin blocks, trimmed, and stained with hematoxylin and eosin. One representative image is provided for each group at two magnifications. A and B) Tumor morphology in the control group; C and D) 0.1 mg/kg Re-diSe; E and F) 1 mg/kg Re-diSe; G and H: 10 mg/kg Re-diSe. Pink staining (eosin) is specific for cell’s cytoplasm and extracellular matrix, while purple staining (hematoxylin) is specific for cell’s nucleus. Yellow arrows indicate necrotic areas and red arrows proliferative ones. Scale bar=500 μm (A, C, E & G) and 50 μm (B, D, F & H).

Discussion

Efficacy. The efficacy of Re-diSe was demonstrated at a dose as low as 0.1 mg/kg for a relatively short duration of administration, with a significant decrease in the tumor weight compared with the controls. Increasing the dose to 1 mg/kg did not decrease the tumor weight but induced an increase in the number of inflammatory cells. At the dose of 10 mg/kg, the decrease of the tumor weights was highly significant compared with the controls. This was confirmed by the histological analysis, which showed a rarefaction of the tumor cells or even a tumor necrosis and an increase in the number of inflammatory cells. The tumor weights did not significantly vary between 1 and 10 mg/kg but the 6/11 embryos without visible tumors were excluded from the statistics. Only measurable tumors in 5/11 embryos were included in the statistical analysis. In any case, at the dose of 10 mg/kg a remarkable anticancer activity was achieved.

Toxicity. There was an increase in the number of deaths of embryos in the treated groups, but the difference was not significant versus controls. The toxicity of Re-diSe was previously evaluated in other in vivo models. The toxicity observed in mice varied according to the mode of administration. A toxicity was noted after intraperitoneal (IP) injections (5). The 60 mg/kg dose was considered as the maximum tolerated dose (MTD) for a four-week daily IP administration, 75 mg/kg was the 50% lethal dose (LD50) dose and 40 mg/kg the LD10. Finally, the 10 mg/kg-treated group showed no sign of toxicity, and this was the no-adverse toxicity level. However, the oral administration of Re-diSe did not induce any sign of toxicity at the daily dose of 10 mg/kg for six weeks, even in combination with a weekly administration of paclitaxel during the last two weeks of treatment (13). Similar results were obtained with the daily dose of 60 mg/kg for 3 weeks (14). The mode of administration in the chicken embryo model is similar to the IP injection as the cells are directly inoculated on the membrane. An oral administration should be less toxic than a parenteral route, with the advantage to allow repeated administrations. The bioavailability of Re-diSe after an oral administration was demonstrated by the high and dose-dependent plasma concentrations of Re (15) as well as the dose-dependent tissue and tumor Re uptake (13, 16).

Inflammation/Immune cell infiltration. We observed a significant increase in the number of inflammatory cells in tumors treated with 1 and 10 mg/kg, but not in the group treated with 0.1 mg/kg, which induced a significant reduction of the tumor weights.

The term of inflammation is usual in histopathology, but microscopic observations without immuno-histochemistry assays cannot distinguish immune cells with pro-inflammatory properties from immune cells with anti-inflammatory properties. Moreover, cells of the tumor microenvironment (TME) are multiple. Lymphocytes, macrophages and dendritic cells are the main immune cells of the TME but tumor-associated stromal cells (TASCs), cancer-associated fibroblasts (CAFs), tumor-associated neutrophils (TANs), tumor-associated endothelial cells (TECs), mesenchymal stromal cells (MSCs) and cancer-associated adipocytes (CAA) also participate to the cancer development (17-19). There is a great heterogeneity between all these cells. In order to understand the immune effects of Re-diSe, we first give an overview on immune suppressive or immune stimulatory cells.

Immune suppressive and immune stimulatory cells. Myeloid cells, comprised macrophages, dendritic cells, monocytes, and granulocytes, represent a major component of the TME with either immune suppressive or immune stimulatory roles (20). They also play an important role in phagocytosis and antigen presentation to T-cells (21).

In the pathological state of activation, mainly induced by a lipid peroxidation, myeloid cells have an immune-suppressive role by inhibiting T cell immunity and the cytolytic function of intra-tumoral natural killer (NK) cells through the production of IL-10 and TGF-β and are defined as “myeloid-derived suppressor cells” (MDSCs) (22, 23). The granulocytic myeloid-derived suppressor cell (G-MDSC) is the most widely distributed subtype in tumors (24).

Tumor-associated macrophages (TAMs) may have pro- or anti-inflammatory properties (25) and their role is primordial in the cancer development (26). The significant incidence of macrophage infiltration is commonly correlated with an unfavorable prognosis, but it could be more relevant to characterize the type of macrophages.

While M2 macrophages exert an immune-suppressive effect, M1 macrophages stimulate inflammation against cancer cells (27). The therapeutic strategy could be to increase the number of M1-like macrophages and to decrease the M2-like macrophages but selectively in the tumor to avoid a dangerous systemic inflammation by M1-like macrophages or an excessive lack of function of M2-like macrophages.

T lymphocytes are the principal component of tumor-infiltrating lymphocytes (TILs). The number of TILs depends on the subtype of cancer (28-30). In breast cancers, the highest level of TILs was observed in triple-negative breast cancer (TNBC) which is the most aggressive subtype (31). However, the role of TILs may differ according to the type of lymphocytes present in the TME. The distinction between the involved lymphocytes could be more important than measuring their total number to evaluate their effects in cancer progression and invasiveness. CD8+ cytotoxic T cells mediate effective anti- tumor immunity and are the most powerful effectors in the anticancer immune response (32). CD4+ helper T cells (Th) may have distinct roles according to their subtypes, either Th1 or Th2 (33, 34). The differentiation of T-helper cells to Th1 and Th2 subsets is of prognostic value (35). CD25+CD4+ regulatory T cells (Tregs) are highly immune-suppressive, expressing FoxP3 (36). The highest levels of FOXP3+ cells are observed in TNBC (28). NK cells exert cytotoxic functions, eradicating tumor cells through cytolytic granules and cooperating with other innate and adaptive immune cells through proinflammatory cytokines (37).

Anti-inflammatory and pro-tumor cells are M2-type macrophages, regulatory T (Treg) cells, polymorphonuclear (PMN) and monocytic (M) MDSCs. They are considered immune-suppressive cells. They have been shown to be significantly increased in the tumor immune microenvironment (TIME (17). Th2 cells have also been linked to tumor promotion and positively correlated with Treg cells (38).

Pro-Inflammatory and anti-tumor cells are M1-type macrophages, CD8+ cytotoxic T cells and Th1 cells. They mediate an effective anti- tumor immunity in advanced cancers and facilitate the recruitment and activation of NK cells against cancer (39).

Cysteine proteases-mediated immune effects of Re-diSe on the polarization of macrophages. It was demonstrated that Re-diSe significantly increased the number of M1-like macrophages and significantly decreased the number of M2-like macrophages (40). To induce the polarization into M1 macrophages, murine Raw 264 or human THP-1 macrophages were previously treated with liposaccharides (LPS), while to induce the polarization into M2, the cells were stimulated with interleukin IL-6. The stimulation with LPS reflects the increased inflammatory situation observed in the TME. The increased number of M1 macrophages induced by Re-diSe could explain the destruction of the tumor cells observed in our study, while the decreased number of M2 macrophages could have counteracted the immune-resistance of the tumor cells. It is also important to note that Re-diSe had no effect on non-prior stimulated macrophages, thus in the absence of inflammation.

The effects of Re-diSe on the polarization of the macrophages were explained by the effects of Re-diSe on the cysteine proteases cathepsins B and S (20). It was demonstrated that Re-diSe decreased the production of cathepsins B and S in macrophages (40) and in cancer cells (41). Cathepsins B and S are over-expressed in cancer cells and in macrophages (42). They are involved in cancer progression and in the immune/inflammation response of the cells of the TME (43). Serum levels of cathepsin S increased with the stage of cancer and are over-expressed at late metastatic stage (44). Inhibition of the activity of cathepsins B and S induced antitumor effects (45-49), and a shift from M2 to M1-like phenotype, with an increased expression of autophagy- and lysosome-associated marker genes, changes in lysosomal activity, fatty acid metabolism, synthesis of pro-inflammatory mediators and reduced adenosine triphosphate (ATP) levels (50).

Re-diSe could have an immunotherapeutic activity in addition to its selective inhibitory effects on cancer cells when cysteine proteases are produced in excess. However, cysteine proteases are necessary for an optimal function of immune cells. The objective should be to normalize the production of cysteine proteases when they are over-expressed and not to decrease them below the optimal level required for a normal function of the immune cells.

Reactive oxygen species (ROS)/Reactive nitrogen species (RNS) mediated-effects of Re-diSe on immune and cancer cells. The binding of the Re atom of the Re-diSe drug with thiol (SH) groups was clearly demonstrated in the cases of N-acetylcysteine and of glutathione (GSH) (40). By this mechanism, Re-diSe will affect the redox status of immune and cancer cells.

Binding of Re to SH groups. The SH groups of redox-active Cys residues have high nucleophilic properties, a high affinity metal binding and the ability to form disulfide bonds with an exchange between reduced dithiol-containing molecules and oxidized disulfide-containing molecules (51). Protein and non-protein thiols are the first targets of ROS (52). Through the binding of Re-diSe with SH groups, the Re-diSe could affect protein oxidation, which commonly occurs at thiol-containing cysteine residues, by preventing disulfide bridge formation (52). There are many SH-rich proteins in cancer cells. For example, Park et al. identified 194 reactive SH-containing proteins in prostate cancer cells (53). It was also observed that cationic Re complexes could covalently bind to SH-rich mitochondrial components (54). The redox status of the immune and cancer cells may thus be modified by the Re-diSe treatment. In cancer cells, there is usually a pro-oxidative status (55). Oxidation-reduction in cancer cells are in favor of cellular oxidation with a more pronounced pro-oxidant status when the stage of the cancer is more advanced (56). MDSCs are also induced by oxidation, as previously described. It is via an oxidative-stress-associated mechanism that apoptotic Treg cells achieved superior suppressor function, inhibiting spontaneous and checkpoint- blockade-induced antitumor immunity (57).

The particular case of GSH. The binding of the Re atom of Re-diSe with GSH may have major consequences. The GSH levels are high in cancer cells to protect them from the oxidative damage induced by anticancer drugs (58). Excess GSH promotes tumor progression, where elevated levels correlate with increased metastasis (59). Depletion of GSH has been proposed as a new strategy against cancer, with ferroptosis as a possible result (60). Ferroptosis could be a mode of Re-diSe-induced death at high doses, resulting in significant Cys depletion (61). At lower doses reaching and respecting the normal values of thiols, Re-diSe could induce favorable immune effects on the polarization of macrophages.

Decreased production of ROS, TGF-β1, VEGF-A, and IGF-1. A dose-dependent decrease in the production of ROS, TGF-β1, VEGF-A, and IGF-1 was observed in MDA-MB-231 breast cancer cells treated with Re-diSe, but not in normal HEK-293 cells (2). The Re-diSe complex was more effective than its di-Se ligand, suggesting that Re plays a critical role in the anticancer activity.

ROS play an important role as signaling messengers in the immune system. They regulate T cell immune response in the TME (62), but also the fate and function of MDSCs (63). Tumor growth is associated with the accumulation of immature myeloid cells (ImC) with significantly higher levels of ROS than ImC isolated from tumor-free animals. They suppress Ag-specific CD8+ T cells via direct cell-cell contact. Inhibition of ROS in ImC completely abrogated the inhibitory effect of these cells on T cells (64). The decreased ROS production by Re-diSe could thus have indirect effects on innate and adaptive immunity.

TGF-β1, VEGF-A, and IGF-1 are redox-dependent factors, greatly involved in cancer progression and immune-suppression. TGF-β regulates infiltration of inflammatory/immune cells and cancer-associated fibroblasts in the TME causing direct changes in tumor cells. Neutralizing TGF-β enhanced CD8+ T-cell- and NK-cell-mediated anti-tumor immune responses (65). A decrease of VEGF increased NK cell mediated lysis of an ovarian cancer cell line and enhanced the cytolytic activity of immune T cells, without toxicity for the immune cells at doses providing cytotoxic effects for the cancer cells (66). Insulin/IGF-1 signaling promotes immunosuppression via the STAT3 pathway (67) and stimulates the expansion of Tregs, increasing their immunosuppressive activity (68).

Through its action on ROS, TGF-β1, VEGF-A, and IGF-1 the Re-diSe could then increase the number of cytotoxic T cells and NK cells, while decreasing MDSCs cells, and the expansion of Tregs, fighting the immune-resistance of cancer cells.

Binding of Re with DNA. The binding of Re to guanine bases was demonstrated via the formation of mono- or bis-adducts with Re-diSe (4). Re was found in the nucleus of cancer cells exposed to Re-diSe (16). Nearly all tricarbonyl(I)-Re complexes form reversible bonds with either adenine or guanine bases, in contrast to the binding of cisplatin with guanine bases which is irreversible (1). However, the mitochondrial DNA could also be a target of Re-diSe. For example, it was shown that the binding of a tricarbonyl(I)-Re complex with doxorubicin diverted the normal accumulation of doxorubicin from the nucleus to mitochondria (69). Moreover, it has been recently shown that mitochondria were a target of third row transition metal-based anticancer complexes (70). The mitochondrial DNA is greatly involved in ROS and RNS productions and the binding of Re with the mitochondrial DNA could be related to its effect on reactive species. Further studies are necessary to confirm this hypothesis.

Tumor necrosis. The role of inflammatory/immune cells in the necrosis of the tumor is highly suggested by the results observed at the dose of 10 mg/kg, with increased inflammation. It is the first time that the efficacy of Re-diSe is correlated with immune cells infiltration with a remarkable tumor necrosis at the dose of 10 mg/kg, which is the main result of this study.

An increased number of M1-like macrophages could be expected as already emphasized, with a decrease in M2-like macrophages. However, via the effects of Re-diSe on multiple pathways, an activation of NK cells and an increased number of cytotoxic cells, combined with a decreased role of MDSCs and Tregs are expected. Re-diSe could then be considered a new immunotherapeutic drug.

In any case, the doses of Re-diSe should be managed to reach and respect the normal levels of GSH, cysteine proteases, ROS and RNS in the immune cells, while inhibiting the growth of cancer cells.

An autophagic type of death of cancer cells was observed with other Re compounds and linked to lysosomal dysfunctions involving cathepsin protease B (71). A paraptosis pathway for cell killing with vacuole formations was also observed in prostate cancer PC3 cells exposed to Re complexes (72).

The mode of cancer cell death induced by Re-diSe has still to be clarified, but the anticancer and immune effects of Re-diSe are demonstrated.

Conclusion

The efficacy of the Re-diSe was clearly demonstrated in this in vivo chicken embryo model with a significant reduction of the tumor weights at the low dose of 0.1 mg/Kg, which is safe. At the dose of 10 mg/kg, high inflammation and huge tumor necrosis were observed, with inflammatory infiltration of the CAM. Remarkably, no visible tumors were observed in more than 50% of the treated embryos at this dose. The immune effects suggested by the high inflammation score may be related to the known role of Re-diSe on the polarization of macrophages, via the decrease on cysteine proteases, and on the adaptive immunity through its indirect anti-oxidant effects.

Footnotes

  • Authors’ Contributions

    Conceptualization: Philippe Collery, Adhikesavan Harikrishnan, Vijay Veena and Chloé Prunier; methodology, Chloé Prunier, Emilien Dosda and Jean Viallet; Synthesis of the Re-diSe drug: Didier Desmaële; validation: Chloé Prunier and Emilien Dosda; investigation: Chloé Prunier; resources: Philippe Collery; writing: Philippe Collery, Chloé Prunier and Didier Desmaële; project administration: Philippe Collery and Didier Desmaële; funding acquisition: Philippe Collery. All Authors have read and agreed to the published version of the manuscript.

  • Conflicts of Interest

    Philippe Collery has been designated as inventor on a patent on “Rhenium complexes and their pharmaceutical use”. He is manager of the Society for the Coordination of Therapeutic Research, which is owner of the Intellectual Property Rights. The other Authors declare that they have no conflict of interest in relation to this study.

  • Funding

    This research received no external funding.

  • Received November 21, 2023.
  • Revision received December 25, 2023.
  • Accepted December 27, 2023.
  • Copyright © 2024 The Author(s). Published by the International Institute of Anticancer Research.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Collery P,
    2. Desmaele D,
    3. Vijaykumar V
    : Design of rhenium compounds in targeted anticancer therapeutics. Curr Pharm Des 25(31): 3306-3322, 2019. DOI: 10.2174/1381612825666190902161400
    OpenUrlCrossRefPubMed
  2. ↵
    1. Collery P,
    2. Veena V,
    3. Harikrishnan A,
    4. Desmaele D
    : The rhenium(I)-diselenoether anticancer drug targets ROS, TGF-β1, VEGF-A, and IGF-1 in an in vitro experimental model of triple-negative breast cancers. Invest New Drugs 37(5): 973-983, 2019. DOI: 10.1007/s10637-019-00727-1
    OpenUrlCrossRefPubMed
  3. ↵
    1. Veena V,
    2. Harikrishnan A,
    3. Lakshmi B,
    4. Khanna S,
    5. Desmaele D,
    6. Collery P
    : A new model applied for evaluating a rhenium-diselenium drug: Breast cancer cells stimulated by cytokines induced from polynuclear cells by LPS. Anticancer Res 40(4): 1915-1920, 2020. DOI: 10.21873/anticanres.14146
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Collery P,
    2. Mohsen A,
    3. Kermagoret A,
    4. Corre S,
    5. Bastian G,
    6. Tomas A,
    7. Wei M,
    8. Santoni F,
    9. Guerra N,
    10. Desmaële D,
    11. d’Angelo J
    : Antitumor activity of a rhenium (I)-diselenoether complex in experimental models of human breast cancer. Invest New Drugs 33(4): 848-860, 2015. DOI: 10.1007/s10637-015-0265-z
    OpenUrlCrossRefPubMed
  5. ↵
    1. Collery P,
    2. Santoni F,
    3. Ciccolini J,
    4. Tran TNN,
    5. Mohsen A,
    6. Desmaele D
    : Dose effect of rhenium (I)-diselenoether as anticancer drug in resistant breast tumor-bearing mice after repeated administrations. Anticancer Res 36(11): 6051-6058, 2016. DOI: 10.21873/anticanres.11194
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Schneider-Stock R,
    2. Ribatti D
    : The CAM assay as an alternative in vivo model for drug testing. Handb Exp Pharmacol 265: 303-323, 2020. DOI: 10.1007/164_2020_375
    OpenUrlCrossRef
    1. Harper K,
    2. Yatsyna A,
    3. Charbonneau M,
    4. Brochu-Gaudreau K,
    5. Perreault A,
    6. Jeldres C,
    7. McDonald PP,
    8. Dubois CM
    : The chicken chorioallantoic membrane tumor assay as a relevant in vivo model to study the impact of hypoxia on tumor progression and metastasis. Cancers (Basel) 13(5): 1093, 2021. DOI: 10.3390/cancers13051093
    OpenUrlCrossRefPubMed
  7. ↵
    1. Miebach L,
    2. Berner J,
    3. Bekeschus S
    : In ovo model in cancer research and tumor immunology. Front Immunol 13: 1006064, 2022. DOI: 10.3389/fimmu.2022.1006064
    OpenUrlCrossRefPubMed
  8. ↵
    1. Dutta S,
    2. Going JJ,
    3. Crumley AB,
    4. Mohammed Z,
    5. Orange C,
    6. Edwards J,
    7. Fullarton GM,
    8. Horgan PG,
    9. McMillan DC
    : The relationship between tumour necrosis, tumour proliferation, local and systemic inflammation, microvessel density and survival in patients undergoing potentially curative resection of oesophageal adenocarcinoma. Br J Cancer 106(4): 702-710, 2012. DOI: 10.1038/bjc.2011.610
    OpenUrlCrossRefPubMed
    1. Fujii T,
    2. Shimada K,
    3. Asai O,
    4. Tanaka N,
    5. Fujimoto K,
    6. Hirao K,
    7. Konishi N
    : Immunohistochemical analysis of inflammatory cells in benign and precancerous lesions and carcinoma of the prostate. Pathobiology 80(3): 119-126, 2013. DOI: 10.1159/000342396
    OpenUrlCrossRefPubMed
  9. ↵
    1. Zhang Y,
    2. Ren H,
    3. Wang L,
    4. Ning Z,
    5. Zhuang Y,
    6. Gan J,
    7. Chen S,
    8. Zhou D,
    9. Zhu H,
    10. Tan D,
    11. Zhang H
    : Clinical impact of tumor-infiltrating inflammatory cells in primary small cell esophageal carcinoma. Int J Mol Sci 15(6): 9718-9734, 2014. DOI: 10.3390/ijms15069718
    OpenUrlCrossRefPubMed
  10. ↵
    1. Klintrup K,
    2. Mäkinen JM,
    3. Kauppila S,
    4. Väre PO,
    5. Melkko J,
    6. Tuominen H,
    7. Tuppurainen K,
    8. Mäkelä J,
    9. Karttunen TJ,
    10. Mäkinen MJ
    : Inflammation and prognosis in colorectal cancer. Eur J Cancer 41(17): 2645-2654, 2005. DOI: 10.1016/j.ejca.2005.07.017
    OpenUrlCrossRefPubMed
  11. ↵
    1. Collery P,
    2. Santoni F,
    3. Mohsen A,
    4. Mignard C,
    5. Desmaele D
    : Negative impact of total body irradiation on the antitumor activity of Rhenium-(I)-diselenoether. Anticancer Res 36(11): 5813-5830, 2016. DOI: 10.21873/anticanres.11165
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Collery P,
    2. Lagadec P,
    3. Krossa I,
    4. Cohen C,
    5. Antomarchi J,
    6. Varlet D,
    7. Lucio M,
    8. Guigonis JM,
    9. Scimeca JC,
    10. Schmid-Antomarchi H,
    11. Schmid-Alliana A
    : Relationship between the oxidative status and the tumor growth in transplanted triple-negative 4T1 breast tumor mice after oral administration of rhenium(I)-diselenoether. J Trace Elem Med Biol 71: 126931, 2022. DOI: 10.1016/J.JTEMB.2022.126931
    OpenUrlCrossRefPubMed
  13. ↵
    1. Collery P,
    2. Michalke B,
    3. Lucio M,
    4. Varlet D,
    5. Guigonis JM,
    6. Scimeca JC,
    7. Schmid-Antomarchi H,
    8. Schmid-Alliana A
    : Plasma rhenium and selenium concentrations after repeated daily oral administration of Rhenium(I)-diselenoether in 4T1 breast tumor-bearing mice. Anticancer Res 43(3): 1017-1023, 2023. DOI: 10.21873/anticanres.16246
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Collery P,
    2. Bastian G,
    3. Santoni F,
    4. Mohsen A,
    5. Wei M,
    6. Collery T,
    7. Tomas A,
    8. Desmaele D,
    9. D’Angelo J
    : Uptake and efflux of rhenium in cells exposed to rhenium diseleno-ether and tissue distribution of rhenium and selenium after rhenium diselenoether treatment in mice. Anticancer Res 34: 1679-1690, 2014.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Mao X,
    2. Xu J,
    3. Wang W,
    4. Liang C,
    5. Hua J,
    6. Liu J,
    7. Zhang B,
    8. Meng Q,
    9. Yu X,
    10. Shi S
    : Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer 20(1): 131, 2021. DOI: 10.1186/s12943-021-01428-1
    OpenUrlCrossRefPubMed
    1. Xu M,
    2. Zhang T,
    3. Xia R,
    4. Wei Y,
    5. Wei X
    : Targeting the tumor stroma for cancer therapy. Mol Cancer 21(1): 208, 2022. DOI: 10.1186/s12943-022-01670-1
    OpenUrlCrossRef
  16. ↵
    1. Bussard KM,
    2. Mutkus L,
    3. Stumpf K,
    4. Gomez-Manzano C,
    5. Marini FC
    : Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res 18(1): 84, 2016. DOI: 10.1186/s13058-016-0740-2
    OpenUrlCrossRefPubMed
  17. ↵
    1. Jahchan NS,
    2. Mujal AM,
    3. Pollack JL,
    4. Binnewies M,
    5. Sriram V,
    6. Reyno L,
    7. Krummel MF
    : Tuning the tumor myeloid microenvironment to fight cancer. Front Immunol 10: 1611, 2019. DOI: 10.3389/fimmu.2019.01611
    OpenUrlCrossRef
  18. ↵
    1. van Vlerken-Ysla L,
    2. Tyurina YY,
    3. Kagan VE,
    4. Gabrilovich DI
    : Functional states of myeloid cells in cancer. Cancer Cell 41(3): 490-504, 2023. DOI: 10.1016/j.ccell.2023.02.009
    OpenUrlCrossRef
  19. ↵
    1. Gabrilovich DI,
    2. Nagaraj S
    : Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9(3): 162-174, 2009. DOI: 10.1038/nri2506
    OpenUrlCrossRefPubMed
  20. ↵
    1. Li H,
    2. Han Y,
    3. Guo Q,
    4. Zhang M,
    5. Cao X
    : Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-β1. J Immunol 182(1): 240-249, 2009. DOI: 10.4049/jimmunol.182.1.240
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Zhao X,
    2. Li Z,
    3. Gu Z
    : A new era: tumor microenvironment in chemoresistance of pancreatic cancer. J Cancer Sci Clin Ther 6(1): 61-86, 2022. DOI: 10.26502/jcsct.5079146
    OpenUrlCrossRefPubMed
  22. ↵
    1. Singh S,
    2. Mehta N,
    3. Lilan J,
    4. Budhthoki MB,
    5. Chao F,
    6. Yong L
    : Initiative action of tumor-associated macrophage during tumor metastasis. Biochim Open 4: 8-18, 2017. DOI: 10.1016/j.biopen.2016.11.002
    OpenUrlCrossRefPubMed
  23. ↵
    1. Jahandideh A,
    2. Yarizadeh M,
    3. Noei-Khesht Masjedi M,
    4. Fatehnejad M,
    5. Jahandideh R,
    6. Soheili R,
    7. Eslami Y,
    8. Zokaei M,
    9. Ahmadvand A,
    10. Ghalamkarpour N,
    11. Kumar Pandey R,
    12. Nabi Afjadi M,
    13. Payandeh Z
    : Macrophage’s role in solid tumors: two edges of a sword. Cancer Cell Int 23(1): 150, 2023. DOI: 10.1186/s12935-023-02999-3
    OpenUrlCrossRefPubMed
  24. ↵
    1. Liu J,
    2. Geng X,
    3. Hou J,
    4. Wu G
    : New insights into M1/M2 macrophages: key modulators in cancer progression. Cancer Cell Int 21(1): 389, 2021. DOI: 10.1186/s12935-021-02089-2
    OpenUrlCrossRefPubMed
  25. ↵
    1. Stanton SE,
    2. Adams S,
    3. Disis ML
    : Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes. JAMA Oncol 2(10): 1354, 2016. DOI: 10.1001/jamaoncol.2016.1061
    OpenUrlCrossRefPubMed
    1. Stanton SE,
    2. Disis ML
    : Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J Immunother Cancer 4: 59, 2016. DOI: 10.1186/s40425-016-0165-6
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Denkert C,
    2. von Minckwitz G,
    3. Darb-Esfahani S,
    4. Lederer B,
    5. Heppner BI,
    6. Weber KE,
    7. Budczies J,
    8. Huober J,
    9. Klauschen F,
    10. Furlanetto J,
    11. Schmitt WD,
    12. Blohmer JU,
    13. Karn T,
    14. Pfitzner BM,
    15. Kümmel S,
    16. Engels K,
    17. Schneeweiss A,
    18. Hartmann A,
    19. Noske A,
    20. Fasching PA,
    21. Jackisch C,
    22. van Mackelenbergh M,
    23. Sinn P,
    24. Schem C,
    25. Hanusch C,
    26. Untch M,
    27. Loibl S
    : Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 19(1): 40-50, 2018. DOI: 10.1016/S1470-2045(17)30904-X
    OpenUrlCrossRefPubMed
  27. ↵
    1. Yu T,
    2. Di G
    : Role of tumor microenvironment in triple-negative breast cancer and its prognostic significance. Chin J Cancer Res 29(3): 237-252, 2017. DOI: 10.21147/j.issn.1000-9604.2017.03.10
    OpenUrlCrossRefPubMed
  28. ↵
    1. Raskov H,
    2. Orhan A,
    3. Christensen JP,
    4. Gögenur I
    : Cytotoxic CD8(+) T cells in cancer and cancer immunotherapy. Br J Cancer 124(2): 359-367, 2021. DOI: 10.1038/s41416-020-01048-4
    OpenUrlCrossRefPubMed
  29. ↵
    1. Boieri M,
    2. Malishkevich A,
    3. Guennoun R,
    4. Marchese E,
    5. Kroon S,
    6. Trerice KE,
    7. Awad M,
    8. Park JH,
    9. Iyer S,
    10. Kreuzer J,
    11. Haas W,
    12. Rivera MN,
    13. Demehri S
    : CD4+ T helper 2 cells suppress breast cancer by inducing terminal differentiation. J Exp Med 219(7): e20201963, 2022. DOI: 10.1084/jem.20201963
    OpenUrlCrossRef
  30. ↵
    1. Johansson M,
    2. Denardo DG,
    3. Coussens LM
    : Polarized immune responses differentially regulate cancer development. Immunol Rev 222: 145-154, 2008. DOI: 10.1111/j.1600-065X.2008.00600.x
    OpenUrlCrossRefPubMed
  31. ↵
    1. Haslund-Vinding J,
    2. Møller JR,
    3. Ziebell M,
    4. Vilhardt F,
    5. Mathiesen T
    : The role of systemic inflammatory cells in meningiomas. Neurosurg Rev 45(2): 1205-1215, 2022. DOI: 10.1007/s10143-021-01642-x
    OpenUrlCrossRefPubMed
  32. ↵
    1. Togashi Y,
    2. Shitara K,
    3. Nishikawa H
    : Regulatory T cells in cancer immunosuppression – implications for anticancer therapy. Nat Rev Clin Oncol 16(6): 356-371, 2019. DOI: 10.1038/s41571-019-0175-7
    OpenUrlCrossRefPubMed
  33. ↵
    1. Paul S,
    2. Lal G
    : The molecular mechanism of natural killer cells function and its importance in cancer immunotherapy. Front Immunol 8: 1124, 2017. DOI: 10.3389/fimmu.2017.01124
    OpenUrlCrossRefPubMed
  34. ↵
    1. Zhou Y,
    2. Tian Q,
    3. Gao H,
    4. Zhu L,
    5. Zhang Y,
    6. Zhang C,
    7. Yang J,
    8. Wang B
    : Immunity and extracellular matrix characteristics of breast cancer subtypes based on identification by T helper cells profiling. Front Immunol 13: 859581, 2022. DOI: 10.3389/fimmu.2022.859581
    OpenUrlCrossRefPubMed
  35. ↵
    1. Braumüller H,
    2. Wieder T,
    3. Brenner E,
    4. Aßmann S,
    5. Hahn M,
    6. Alkhaled M,
    7. Schilbach K,
    8. Essmann F,
    9. Kneilling M,
    10. Griessinger C,
    11. Ranta F,
    12. Ullrich S,
    13. Mocikat R,
    14. Braungart K,
    15. Mehra T,
    16. Fehrenbacher B,
    17. Berdel J,
    18. Niessner H,
    19. Meier F,
    20. van den Broek M,
    21. Häring HU,
    22. Handgretinger R,
    23. Quintanilla-Martinez L,
    24. Fend F,
    25. Pesic M,
    26. Bauer J,
    27. Zender L,
    28. Schaller M,
    29. Schulze-Osthoff K,
    30. Röcken M
    : T-helper-1-cell cytokines drive cancer into senescence. Nature 494(7437): 361-365, 2013. DOI: 10.1038/nature11824
    OpenUrlCrossRefPubMed
  36. ↵
    1. Collery P,
    2. Desmaële D,
    3. Harikrishnan A,
    4. Veena V
    : Remarkable effects of a Rhenium(I)-diselenoether drug on the production of Cathepsins B and S by macrophages and their polarizations. Curr Pharm Des 29(30): 2396-2407, 2023. DOI: 10.2174/0113816128268963231013074433
    OpenUrlCrossRefPubMed
  37. ↵
    1. Collery P,
    2. Veena V,
    3. Desmaële D,
    4. Harikrishnan A,
    5. Lakshmi B
    : Effects of Rhenium(I)-diselenoether and of its diselenide ligand on the production of cathepsins B and S by MDA-MB231 breast malignant cells. Anticancer Res 41(12): 5997-6001, 2021. DOI: 10.21873/anticanres.15418
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Verma S,
    2. Dixit R,
    3. Pandey KC
    : Cysteine proteases: Modes of activation and future prospects as pharmacological targets. Front Pharmacol 7: 107, 2016. DOI: 10.3389/fphar.2016.00107
    OpenUrlCrossRefPubMed
  39. ↵
    1. Joyce JA,
    2. Baruch A,
    3. Chehade K,
    4. Meyer-Morse N,
    5. Giraudo E,
    6. Tsai FY,
    7. Greenbaum DC,
    8. Hager JH,
    9. Bogyo M,
    10. Hanahan D
    : Cathepsin cysteine protease are effectors of invasive growth and angiogenesis during multistage tumorigenesis. Cancer Cell 5: 443-453, 2004.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Liu WL,
    2. Liu D,
    3. Cheng K,
    4. Liu YJ,
    5. Xing S,
    6. Chi PD,
    7. Liu XH,
    8. Xue N,
    9. Lai YZ,
    10. Guo L,
    11. Zhang G
    : Evaluating the diagnostic and prognostic value of circulating cathepsin S in gastric cancer. Oncotarget 7(19): 28124-28138, 2016. DOI: 10.18632/oncotarget.8582
    OpenUrlCrossRefPubMed
  41. ↵
    1. Rudzińska M,
    2. Parodi A,
    3. Soond SM,
    4. Vinarov AZ,
    5. Korolev DO,
    6. Morozov AO,
    7. Daglioglu C,
    8. Tutar Y,
    9. Zamyatnin AA Jr.
    : The role of cysteine cathepsins in cancer progression and drug resistance. Int J Mol Sci 20(14): 3602, 2019. DOI: 10.3390/ijms20143602
    OpenUrlCrossRefPubMed
    1. Pogorzelska A,
    2. Żołnowska B,
    3. Bartoszewski R
    : Cysteine cathepsins as a prospective target for anticancer therapies – current progress and prospects. Biochimie 151: 85-106, 2018. DOI: 10.1016/j.biochi.2018.05.023
    OpenUrlCrossRefPubMed
    1. Li YY,
    2. Fang J,
    3. Ao GZ
    : Cathepsin B and L inhibitors: a patent review (2010 - present). Expert Opin Ther Pat 27(6): 643-656, 2017. DOI: 10.1080/13543776.2017.1272572
    OpenUrlCrossRefPubMed
    1. Fuchs N,
    2. Meta M,
    3. Schuppan D,
    4. Nuhn L,
    5. Schirmeister T
    : Novel opportunities for Cathepsin S inhibitors in cancer immunotherapy by nanocarrier-mediated delivery. Cells 9(9): 2021, 2020. DOI: 10.3390/cells9092021
    OpenUrlCrossRefPubMed
  42. ↵
    1. Kos J,
    2. Mitrović A,
    3. Mirković B
    : The current stage of cathepsin B inhibitors as potential anticancer agents. Future Med Chem 6(11): 1355-1371, 2014. DOI: 10.4155/fmc.14.73
    OpenUrlCrossRefPubMed
  43. ↵
    1. Oelschlaegel D,
    2. Weiss Sadan T,
    3. Salpeter S,
    4. Krug S,
    5. Blum G,
    6. Schmitz W,
    7. Schulze A,
    8. Michl P
    : Cathepsin inhibition modulates metabolism and polarization of tumor-associated macrophages. Cancers (Basel) 12(9): 2579, 2020. DOI: 10.3390/cancers12092579
    OpenUrlCrossRefPubMed
  44. ↵
    1. Poole LB
    : The basics of thiols and cysteines in redox biology and chemistry. Free Radic Biol Med 80: 148-157, 2015. DOI: 10.1016/j.freeradbiomed.2014.11.013
    OpenUrlCrossRefPubMed
  45. ↵
    1. Zhou J,
    2. Li XY,
    3. Liu YJ,
    4. Feng J,
    5. Wu Y,
    6. Shen HM,
    7. Lu GD
    : Full-coverage regulations of autophagy by ROS: from induction to maturation. Autophagy 18(6): 1240-1255, 2022. DOI: 10.1080/15548627.2021.1984656
    OpenUrlCrossRefPubMed
  46. ↵
    1. Park EM,
    2. Choi KS,
    3. Park SY,
    4. Kong ES,
    5. Zu K,
    6. Wu Y,
    7. Zhang H,
    8. Park C,
    9. Ip YM
    : A display thiol-proteomics approach to characterize global redox modification of proteins by selenium: Implications for the anticancer action of selenium. Cancer Genomics Proteomics 2: 25-35, 2005.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. Amoroso AJ,
    2. Arthur RJ,
    3. Coogan MP,
    4. Court JB,
    5. Fernández-Moreira V,
    6. Hayes AJ,
    7. Lloyd D,
    8. Millet C,
    9. Pope SJA
    : 3-Chloromethylpyridyl bipyridine fac-tricarbonyl rhenium: a thiol-reactive luminophore for fluorescence microscopy accumulates in mitochondria. New J Chem 32(7): 1097, 2008. DOI: 10.1039/b802215a
    OpenUrlCrossRef
  48. ↵
    1. Collery P
    : Strategies for the development of selenium-based anticancer drugs. J Trace Elem Med Biol 50: 498-507, 2018. DOI: 10.1016/j.jtemb.2018.02.024
    OpenUrlCrossRefPubMed
  49. ↵
    1. Kruk J,
    2. Aboul-Enein HY
    : Reactive oxygen and nitrogen species in carcinogenesis: implications of oxidative stress on the progression and development of several cancer types. Mini Rev Med Chem 17(11): 904-919, 2017. DOI: 10.2174/1389557517666170228115324
    OpenUrlCrossRefPubMed
  50. ↵
    1. Maj T,
    2. Wang W,
    3. Crespo J,
    4. Zhang H,
    5. Wang W,
    6. Wei S,
    7. Zhao L,
    8. Vatan L,
    9. Shao I,
    10. Szeliga W,
    11. Lyssiotis C,
    12. Liu JR,
    13. Kryczek I,
    14. Zou W
    : Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol 18(12): 1332-1341, 2017. DOI: 10.1038/ni.3868
    OpenUrlCrossRefPubMed
  51. ↵
    1. Marini HR,
    2. Facchini BA,
    3. di Francia R,
    4. Freni J,
    5. Puzzolo D,
    6. Montella L,
    7. Facchini G,
    8. Ottaiano A,
    9. Berretta M,
    10. Minutoli L
    : Glutathione: Lights and shadows in cancer patients. Biomedicines 11(8): 2226, 2023. DOI: 10.3390/biomedicines11082226
    OpenUrlCrossRefPubMed
  52. ↵
    1. Bansal A,
    2. Simon MC
    : Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol 217(7): 2291-2298, 2018. DOI: 10.1083/jcb.201804161
    OpenUrlAbstract/FREE Full Text
  53. ↵
    1. Niu B,
    2. Liao K,
    3. Zhou Y,
    4. Wen T,
    5. Quan G,
    6. Pan X,
    7. Wu C
    : Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials 277: 121110, 2021. DOI: 10.1016/j.biomaterials.2021.121110
    OpenUrlCrossRefPubMed
  54. ↵
    1. Mou Y,
    2. Wang J,
    3. Wu J,
    4. He D,
    5. Zhang C,
    6. Duan C,
    7. Li B
    : Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol 12(1): 34, 2019. DOI: 10.1186/s13045-019-0720-y
    OpenUrlCrossRefPubMed
  55. ↵
    1. Chen X,
    2. Song M,
    3. Zhang B,
    4. Zhang Y
    : Reactive oxygen species regulate T cell immune response in the tumor microenvironment. Oxid Med Cell Longev 2016: 1580967, 2016. DOI: 10.1155/2016/1580967
    OpenUrlCrossRefPubMed
  56. ↵
    1. Ohl K,
    2. Tenbrock K
    : Reactive oxygen species as regulators of MDSC-mediated immune suppression. Front Immunol 9: 2499, 2018. DOI: 10.3389/fimmu.2018.02499
    OpenUrlCrossRefPubMed
  57. ↵
    1. Kusmartsev S,
    2. Nefedova Y,
    3. Yoder D,
    4. Gabrilovich DI
    : Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol 172(2): 989-999, 2004. DOI: 10.4049/jimmunol.172.2.989
    OpenUrlAbstract/FREE Full Text
  58. ↵
    1. Yang L,
    2. Pang Y,
    3. Moses HL
    : TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol 31(6): 220-227, 2010. DOI: 10.1016/j.it.2010.04.002
    OpenUrlCrossRefPubMed
  59. ↵
    1. Nair D,
    2. Rådestad E,
    3. Khalkar P,
    4. Diaz-Argelich N,
    5. Schröder A,
    6. Klynning C,
    7. Ungerstedt J,
    8. Uhlin M,
    9. Fernandes AP
    : Methylseleninic acid sensitizes ovarian cancer cells to T-cell mediated killing by decreasing PDL1 and VEGF levels. Front Oncol 8: 407, 2018. DOI: 10.3389/fonc.2018.00407
    OpenUrlCrossRefPubMed
  60. ↵
    1. Salminen A,
    2. Kaarniranta K,
    3. Kauppinen A
    : Insulin/IGF-1 signaling promotes immunosuppression via the STAT3 pathway: impact on the aging process and age-related diseases. Inflamm Res 70(10-12): 1043-1061, 2021. DOI: 10.1007/s00011-021-01498-3
    OpenUrlCrossRefPubMed
  61. ↵
    1. Bilbao D,
    2. Luciani L,
    3. Johannesson B,
    4. Piszczek A,
    5. Rosenthal N
    : Insulin-like growth factor-1 stimulates regulatory T cells and suppresses autoimmune disease. EMBO Mol Med 6(11): 1423-1435, 2014. DOI: 10.15252/emmm.201303376
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Imstepf S,
    2. Pierroz V,
    3. Rubbiani R,
    4. Felber M,
    5. Fox T,
    6. Gasser G,
    7. Alberto R
    : Organometallic rhenium complexes divert doxorubicin to the mitochondria. Angew Chem Int Ed 55(8): 2792-2795, 2016. DOI: 10.1002/anie.201511432
    OpenUrlCrossRef
  63. ↵
    1. Olelewe C,
    2. Awuah SG
    : Mitochondria as a target of third row transition metal-based anticancer complexes. Curr Opin Chem Biol 72: 102235, 2023. DOI: 10.1016/j.cbpa.2022.102235
    OpenUrlCrossRefPubMed
  64. ↵
    1. He L,
    2. Pan ZY,
    3. Qin WW,
    4. Li Y,
    5. Tan CP,
    6. Mao ZW
    : Impairment of the autophagy-related lysosomal degradation pathway by an anticancer rhenium(i) complex. Dalton Trans 48(13): 4398-4404, 2019. DOI: 10.1039/c9dt00322c
    OpenUrlCrossRefPubMed
  65. ↵
    1. Enslin LE,
    2. Purkait K,
    3. Pozza MD,
    4. Saubamea B,
    5. Mesdom P,
    6. Visser HG,
    7. Gasser G,
    8. Schutte-Smith M
    : Rhenium(I) tricarbonyl complexes of 1,10-phenanthroline derivatives with unexpectedly high cytotoxicity. Inorg Chem 62(31): 12237-12251, 2023. DOI: 10.1021/acs.inorgchem.3c00730
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 44 (3)
Anticancer Research
Vol. 44, Issue 3
March 2024
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Effectiveness of Rhenium(I)-diselenoether Low Doses in a Triple-negative Breast Cancer Chicken Embryo Model
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
3 + 5 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Effectiveness of Rhenium(I)-diselenoether Low Doses in a Triple-negative Breast Cancer Chicken Embryo Model
PHILIPPE COLLERY, CHLOE PRUNIER, EMILIEN DOSDA, JEAN VIALLET, ADHIKESAVAN HARIKRISHNAN, VIJAY VEENA, DIDIER DESMAELE
Anticancer Research Mar 2024, 44 (3) 941-951; DOI: 10.21873/anticanres.16889

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Effectiveness of Rhenium(I)-diselenoether Low Doses in a Triple-negative Breast Cancer Chicken Embryo Model
PHILIPPE COLLERY, CHLOE PRUNIER, EMILIEN DOSDA, JEAN VIALLET, ADHIKESAVAN HARIKRISHNAN, VIJAY VEENA, DIDIER DESMAELE
Anticancer Research Mar 2024, 44 (3) 941-951; DOI: 10.21873/anticanres.16889
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Impact of Rhenium(I)-Diselenoether Compound on Cell Adhesion, Migration, Invasion Capacities, and MMP-2 Release in MDA-MB-231 Breast Cancer Cells
  • Google Scholar

More in this TOC Section

  • Simultaneous Targeting of Multiple Hallmarks of Cancer With Recombinant Methioninase, Rapamycin and Chloroquine Is Specific and Synergistic to MiaPaCa-2 Pancreatic-Cancer Cells in Contrast to Hs-27 Normal Fibroblasts
  • Contribution of Xeroderma Pigmentosum Complementation Group C Genotypes to Colorectal Cancer in Taiwanese
  • Phenotypic Alteration and Suppression of Cytotoxicity of Decidual NK Cells After Co-culturing With Different Trophoblastic Cell Lines
Show more Experimental Studies

Similar Articles

Keywords

  • Rhenium
  • selenium
  • Breast cancer
  • inflammation
  • dose-effect
  • chicken embryo model
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire